1
|
Li K, Xie F, Xiong Y, Jiang J, Huang B. Progress in the application of molecular imaging technology in immunological tolerance and immune metabolism visualization research. Front Immunol 2025; 16:1583228. [PMID: 40236707 PMCID: PMC11996769 DOI: 10.3389/fimmu.2025.1583228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Immunological tolerance and immune metabolism play crucial roles in maintaining immune homeostasis and the immune response to diseases. The advancement of molecular imaging technologies, particularly optical molecular imaging, nuclear medicine imaging, and magnetic resonance imaging, has led to a significant progress in the visualization of immune tolerance and immune metabolism. Molecular imaging technologies enable real-time monitoring and analysis of dynamic changes in immune tolerance mechanisms and immune metabolism in living organisms, allowing the development of new strategies for early disease diagnosis, targeted therapy, and immunotherapy. This article reviews the latest advancements in the application of molecular imaging technologies in the fields of immunological tolerance and immune metabolism, with a focus on their applications in the regulation of immune tolerance regulation, immune metabolism, and immunotherapy.
Collapse
Affiliation(s)
- Kailang Li
- Department of Radiology, The Third People’s Hospital In Xindu District of Chengdu, Chengdu, China
| | - Fang Xie
- Department of Oncology, The Second People’s Hospital of Yibin, Yibin, China
| | - Yongfu Xiong
- Department of General Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jin Jiang
- Department of Radiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Bifan Huang
- Department of Radiology, The Third People’s Hospital In Xindu District of Chengdu, Chengdu, China
| |
Collapse
|
2
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
3
|
MRI Contrast Agents in Glycobiology. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238297. [PMID: 36500389 PMCID: PMC9735696 DOI: 10.3390/molecules27238297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022]
Abstract
Molecular recognition involving glycoprotein-mediated interactions is ubiquitous in both normal and pathological natural processes. Therefore, visualization of these interactions and the extent of expression of the sugars is a challenge in medical diagnosis, monitoring of therapy, and drug design. Here, we review the literature on the development and validation of probes for magnetic resonance imaging using carbohydrates either as targeting vectors or as a target. Lectins are important targeting vectors for carbohydrate end groups, whereas selectins, the asialoglycoprotein receptor, sialic acid end groups, hyaluronic acid, and glycated serum and hemoglobin are interesting carbohydrate targets.
Collapse
|
4
|
Li Y, Li T, Chen H, Wang L, Xia Y, Zhang L, Xie Y, Li J, Luo C, Xu Y, Liu Y, Tan W. Engineering AND-Gate Aptamer-Signal Base Conjugates for Targeted Magnetic Resonance Molecular Imaging of Metastatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:17032-17041. [PMID: 35410471 DOI: 10.1021/acsami.1c24048] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In vivo noninvasive molecular imaging requires precise recognition and in situ, real-time imaging of specific cellular and molecular signatures at the site of interest. However, this is often hindered by issues of current imaging probes relating to either the lack of active recognition or the overall nonspecific mechanism of action. Here, we present an aptamer-signal base conjugate (ApSC) concept to engineer AND-gate molecular tools for tumor-targeted molecular imaging. Superior to conventional synthetic methods for imaging probes, our design enables programmable and precise conjugation between recognition and signaling units in a modular synthesis manner with high fidelity for both the conjugating chemistry and binding affinity to the molecular target. Moreover, this design is endowed with simultaneous multivariate activation that readily adapts to tumor microenvironments for signal output, thus providing improved imaging specificity and sensitivity. Such a concept has been successfully shown in magnetic resonance imaging (MRI), the modality of choice for in vivo noninvasive molecular imaging. The engineered ApSC can produce amplified MR signals only after activation by the unique metabolism and dysregulation of redox balance in cancer. In mouse models of xenograft and metastatic breast cancer, the AND-gate molecular MRI probe elicits high imaging contrast in primary tumors and micrometastases. This study promises to provide synthetically accessible scaffolds that can be extended to a large library of advanced molecular imaging tools with varied imaging modalities and mechanisms of action for preventative, predictive, and personalized medicine.
Collapse
Affiliation(s)
- Yazhou Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Ting Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Hong Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yinghao Xia
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Lili Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yuqi Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Jili Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Can Luo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yingjie Xu
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
5
|
Saad MA, Xavierselvan M, Sharif HA, Selfridge S, Pawle R, Varvares M, Mallidi S, Hasan T. Dual Function Antibody Conjugates for Multimodal Imaging and Photoimmunotherapy of Cancer Cells. Photochem Photobiol 2022; 98:220-231. [PMID: 34379796 PMCID: PMC10038131 DOI: 10.1111/php.13501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/08/2021] [Indexed: 11/29/2022]
Abstract
Precision imaging, utilizing molecular targeted agents, is an important tool in cancer diagnostics and guiding therapies. While there are limitations associated with single mode imaging probes, multimodal molecular imaging probes enabling target visualization through complementary imaging technologies provides an attractive alternative. However, there are several challenges associated with designing molecular probes carrying contrast agents for complementary multimodal imaging. Here, we propose a dual function antibody conjugate (DFAC) comprising an FDA approved photosensitizer Benzoporphyrin derivative (BPD) and a naphthalocyanine-based photoacoustic dye (SiNc(OH)) for multimodal infrared (IR) imaging. While fluorescence imaging, through BPD, provides sensitivity, complementing it with photoacoustic imaging, through SiNc(OH), provides a depth-resolved spatial resolution much beyond the optical diffusion limits of fluorescence measurements. Through a series of in vitro experiments, we demonstrate the development and utilization of DFACs for multimodal imaging and photodynamic treatment of squamous cell carcinoma (A431) cell line. The proposed DFACs have potential use in precision imaging applications such as guiding tumor resection surgeries and photodynamic treatment of residual microscopic disease thereby minimizing local recurrence. The data demonstrated in this study merits further investigation for its preclinical and clinical translation.
Collapse
Affiliation(s)
- Mohammad A. Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Science and Technology Center, Tufts University, Medford, MA
| | | | | | | | - Mark Varvares
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, The Massachusetts Eye and Ear, Boston, MA
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Biomedical Engineering, Science and Technology Center, Tufts University, Medford, MA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
6
|
Mitrou A, Feng X, Khan A, Yaroslavsky AN. Feasibility of dual-contrast fluorescence imaging of pathological breast tissues. JOURNAL OF BIOPHOTONICS 2021; 14:e202100007. [PMID: 34010507 DOI: 10.1002/jbio.202100007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/23/2021] [Accepted: 05/18/2021] [Indexed: 06/12/2023]
Abstract
The combination of intravital dye, methylene blue (MB), with molecular cancer marker, pH low insertion peptide (pHLIP) conjugated with fluorescent Alexa532 (Alexa532-pHLIP), was evaluated for enhancing contrast of pathological breast tissue ex vivo. Fresh, thick breast specimens were stained sequentially with Alexa532-pHLIP and aqueous MB and imaged using dual-channel fluorescence microscopy. MB and Alexa532-pHLIP accumulated in the nuclei and cytoplasm of cancer cells, respectively. MB also stained nuclei of normal cells. Some Alexa532-pHLIP fluorescence emission was detected from connective tissue and benign cell membranes. Overall, Alexa532-pHLIP showed high affinity to cancer, while MB highlighted tissue morphology. The results indicate that MB and Alexa532-pHLIP provide complementary information and show promise for the detection of breast cancer.
Collapse
Affiliation(s)
- Androniki Mitrou
- Advanced Biophotonics Laboratory, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Xin Feng
- Advanced Biophotonics Laboratory, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Ashraf Khan
- Department of Pathology, University of Massachusetts Medical School-Baystate, Springfield, Massachusetts, USA
| | - Anna N Yaroslavsky
- Advanced Biophotonics Laboratory, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| |
Collapse
|
7
|
The Protein Toxins Ricin and Shiga Toxin as Tools to Explore Cellular Mechanisms of Internalization and Intracellular Transport. Toxins (Basel) 2021; 13:toxins13060377. [PMID: 34070659 PMCID: PMC8227415 DOI: 10.3390/toxins13060377] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/22/2021] [Indexed: 12/18/2022] Open
Abstract
Protein toxins secreted by bacteria and found in plants can be threats to human health. However, their extreme toxicity can also be exploited in different ways, e.g., to produce hybrid toxins directed against cancer cells and to study transport mechanisms in cells. Investigations during the last decades have shown how powerful these molecules are as tools in cell biological research. Here, we first present a partly historical overview, with emphasis on Shiga toxin and ricin, of how such toxins have been used to characterize processes and proteins of importance for their trafficking. In the second half of the article, we describe how one can now use toxins to investigate the role of lipid classes for intracellular transport. In recent years, it has become possible to quantify hundreds of lipid species using mass spectrometry analysis. Thus, it is also now possible to explore the importance of lipid species in intracellular transport. The detailed analyses of changes in lipids seen under conditions of inhibited toxin transport reveal previously unknown connections between syntheses of lipid classes and demonstrate the ability of cells to compensate under given conditions.
Collapse
|
8
|
Marasini R, Rayamajhi S, Moreno-Sanchez A, Aryal S. Iron(iii) chelated paramagnetic polymeric nanoparticle formulation as a next-generation T1-weighted MRI contrast agent. RSC Adv 2021; 11:32216-32226. [PMID: 35495502 PMCID: PMC9041822 DOI: 10.1039/d1ra05544e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/18/2021] [Indexed: 12/18/2022] Open
Abstract
In pursuit of safer alternatives to Gd-based MRI contrast agents due to its toxicity and organ deposition, herein, we developed a safer and efficient clinically relevant iron(iii) chelated polymeric nanoparticle as a T1-weighted MRI contrast agent.
Collapse
Affiliation(s)
- Ramesh Marasini
- Department of Chemistry, College of Arts and Sciences, Kansas State University, Manhattan, KS 66506, USA
- Nanotechnology Innovation Center of Kansas State (NICKS), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Sagar Rayamajhi
- Department of Chemistry, College of Arts and Sciences, Kansas State University, Manhattan, KS 66506, USA
- Nanotechnology Innovation Center of Kansas State (NICKS), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Anthony Moreno-Sanchez
- Department of Chemistry, College of Arts and Sciences, Kansas State University, Manhattan, KS 66506, USA
- Nanotechnology Innovation Center of Kansas State (NICKS), Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, The University of Texas, Tyler, TX 75799, USA
| |
Collapse
|
9
|
Covington MF, Schwarz SW, Hoffman JM. The Regulatory Process for Imaging Agents and Devices. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00049-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
10
|
Katzenellenbogen JA. The quest for improving the management of breast cancer by functional imaging: The discovery and development of 16α-[ 18F]fluoroestradiol (FES), a PET radiotracer for the estrogen receptor, a historical review. Nucl Med Biol 2021; 92:24-37. [PMID: 32229068 PMCID: PMC7442693 DOI: 10.1016/j.nucmedbio.2020.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/16/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION 16α-[18F]Fluoroestradiol (FES), a PET radiotracer for the estrogen receptor (ER) in breast cancer, was the first receptor-targeted PET radiotracer for oncology and is continuing to prove its value in clinical research, antiestrogen development, and breast cancer care. The story of its conception, design, evaluation and use in clinical studies parallels the evolution of the whole field of receptor-targeted radiotracers, one greatly influenced by the research and intellectual contributions of William C. Eckelman. METHODS AND RESULTS The development of methods for efficient production of fluorine-18, for conversion of [18F]fluoride ion into chemically reactive form, and for its rapid and efficient incorporation into suitable estrogen precursor molecules at high molar activity, were all methodological underpinnings required for the preparation of FES. FES binds to ER with very high affinity, and its in vivo uptake by ER-dependent target tissues in animal models was efficient and selective, findings that preceded its use for PET imaging in patients with breast cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Comparisons between ER levels measured by FES-PET imaging of breast tumors with tissue-specimen ER quantification by IHC and other methods show that imaging provided improved prediction of benefit from endocrine therapies. Serial imaging of ER by FES-PET, before and after dosing patients with antiestrogens, is used to determine the efficacious dose for established antiestrogens and to facilitate clinical development of new ER antagonists. Beyond FES imaging, PET-based hormone challenge tests, which evaluate the functional status of ER by monitoring rapid changes in tumor metabolic or transcriptional activity after a brief estrogen challenge, provide highly sensitive and selective predictions of whether or not there will be a favorable response to endocrine therapies. There is sufficient interest in the clinical applications of FES that FDA approval is being sought for its wider use in breast cancer. CONCLUSIONS FES was the first PET probe for a receptor in cancer, and its development and clinical applications in breast cancer parallel the conceptual evolution of the whole field of receptor-binding radiotracers.
Collapse
Affiliation(s)
- John A Katzenellenbogen
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| |
Collapse
|
11
|
Mankoff DA. PET Imaging in Cancer Clinical Trials. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
12
|
Katzenellenbogen JA, Kilbourn MR, Coenen HH, Volkert W. William C. Eckelman: An anchor of stability with a quiet voice that nurtured a new field. Nucl Med Biol 2021; 92:2-4. [PMID: 33422387 DOI: 10.1016/j.nucmedbio.2020.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/23/2020] [Indexed: 11/25/2022]
Affiliation(s)
| | - Michael R Kilbourn
- Department of Radiology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| | - Heinz H Coenen
- Institute of Neuroscience and Medicine, INM-5: Nuclear Chemistry, Research Centre Juelich, 52425 Juelich, Germany.
| | - Wynn Volkert
- Department of Radiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
13
|
van Dongen GAMS, Beaino W, Windhorst AD, Zwezerijnen GJC, Oprea-Lager DE, Hendrikse NH, van Kuijk C, Boellaard R, Huisman MC, Vugts DJ. The Role of 89Zr-Immuno-PET in Navigating and Derisking the Development of Biopharmaceuticals. J Nucl Med 2020; 62:438-445. [PMID: 33277395 DOI: 10.2967/jnumed.119.239558] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022] Open
Abstract
The identification of molecular drivers of disease and the compelling rise of biotherapeutics have impacted clinical care but have also come with challenges. Such therapeutics include peptides, monoclonal antibodies, antibody fragments and nontraditional binding scaffolds, activatable antibodies, bispecific antibodies, immunocytokines, antibody-drug conjugates, enzymes, polynucleotides, and therapeutic cells, as well as alternative drug carriers such as nanoparticles. Drug development is expensive, attrition rates are high, and efficacy rates are lower than desired. Almost all these drugs, which in general have a long residence time in the body, can stably be labeled with 89Zr for whole-body PET imaging and quantification. Although not restricted to monoclonal antibodies, this approach is called 89Zr-immuno-PET. This review summarizes the state of the art of the technical aspects of 89Zr-immuno-PET and illustrates why it has potential for steering the design, development, and application of biologic drugs. Appealing showcases are discussed to illustrate what can be learned with this emerging technology during preclinical and especially clinical studies about biologic drug formats and disease targets. In addition, an overview of ongoing and completed clinical trials is provided. Although 89Zr-immuno-PET is a young tool in drug development, its application is rapidly expanding, with first clinical experiences giving insight on why certain drug-target combinations might have better perspectives than others.
Collapse
Affiliation(s)
- Guus A M S van Dongen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wissam Beaino
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - N Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Cornelis van Kuijk
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marc C Huisman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle J Vugts
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Kurland BF, Wiggins JR, Coche A, Fontan C, Bouvet Y, Webner P, Divgi C, Linden HM. Whole-Body Characterization of Estrogen Receptor Status in Metastatic Breast Cancer with 16α-18F-Fluoro-17β-Estradiol Positron Emission Tomography: Meta-Analysis and Recommendations for Integration into Clinical Applications. Oncologist 2020; 25:835-844. [PMID: 32374053 PMCID: PMC7543360 DOI: 10.1634/theoncologist.2019-0967] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/02/2020] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor (ER) status by immunohistochemistry (IHC) of cancer tissue is currently used to direct endocrine therapy in breast cancer. Positron emission tomography (PET) with 16α-18F-fluoro-17β-estradiol (18 F-FES) noninvasively characterizes ER ligand-binding function of breast cancer lesions. Concordance of imaging and tissue assays should be established for 18 F-FES PET to be an alternative or complement to tissue biopsy for metastatic lesions. We conducted a meta-analysis of published results comparing 18 F-FES PET and tissue assays of ER status in patients with breast cancer. PubMed and EMBASE were searched for English-language manuscripts with at least 10 patients and low overall risk of bias. Thresholds for imaging and tissue classification could differ between studies but had to be clearly stated. We used hierarchical summary receiver-operating characteristic curve models for the meta-analysis. The primary analysis included 113 nonbreast lesions from 4 studies; an expanded analysis included 327 total lesions from 11 studies. Treating IHC results as the reference standard, sensitivity was 0.78 (95% confidence region 0.65-0.88) and specificity 0.98 (0.65-1.00) for the primary analysis of nonbreast lesions. In the expanded analysis including non-IHC tissue assays and all lesion sites, sensitivity was 0.81 (0.73-0.87) and specificity 0.86 (0.68-0.94). These results suggest that 18 F-FES PET is useful for characterization of ER status of metastatic breast cancer lesions. We also review current best practices for conducting 18 F-FES PET scans. This imaging assay has potential to improve clinically relevant outcomes for patients with (historically) ER-positive metastatic breast cancer, including those with brain metastases and/or lobular histology. IMPLICATIONS FOR PRACTICE: 16α-18F-fluoro-17β-estradiol positron emission tomography (18 F-FES PET) imaging assesses estrogen receptor status in breast cancer in vivo. This work reviews the sensitivity and specificity of 18 F-FES PET in a meta-analysis with reference tissue assays and discusses best practices for use of the tracer as an imaging biomarker. 18 F-FES PET could enhance breast cancer diagnosis and staging as well as aid in therapy selection for patients with metastatic disease. Tissue sampling limitations, intrapatient heterogeneity, and temporal changes in molecular markers make it likely that 18 F-FES PET will complement existing assays when clinically available in the near future.
Collapse
Affiliation(s)
| | - Jay R. Wiggins
- Merlin Biomedical Consulting, LLCHendersonvilleNorth CarolinaUSA
| | | | | | | | | | | | | |
Collapse
|
15
|
Wijetunga I, McVeigh LE, Charalambous A, Antanaviciute A, Carr IM, Nair A, Prasad KR, Ingram N, Coletta PL. Translating Biomarkers of Cholangiocarcinoma for Theranosis: A Systematic Review. Cancers (Basel) 2020; 12:E2817. [PMID: 33007872 PMCID: PMC7601719 DOI: 10.3390/cancers12102817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 12/16/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a rare disease with poor outcomes and limited research efforts into novel treatment options. A systematic review of CCA biomarkers was undertaken to identify promising biomarkers that may be used for theranosis (therapy and diagnosis). MEDLINE/EMBASE databases (1996-2019) were systematically searched using two strategies to identify biomarker studies of CCA. The PANTHER Go-Slim classification system and STRING network version 11.0 were used to interrogate the identified biomarkers. The TArget Selection Criteria for Theranosis (TASC-T) score was used to rank identified proteins as potential targetable biomarkers for theranosis. The following proteins scored the highest, CA9, CLDN18, TNC, MMP9, and EGFR, and they were evaluated in detail. None of these biomarkers had high sensitivity or specificity for CCA but have potential for theranosis. This review is unique in that it describes the process of selecting suitable markers for theranosis, which is also applicable to other diseases. This has highlighted existing validated markers of CCA that can be used for active tumor targeting for the future development of targeted theranostic delivery systems. It also emphasizes the relevance of bioinformatics in aiding the search for validated biomarkers that could be repurposed for theranosis.
Collapse
Affiliation(s)
- Imeshi Wijetunga
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - Laura E. McVeigh
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - Antonia Charalambous
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - Agne Antanaviciute
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - Ian M. Carr
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - Amit Nair
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - K. Raj Prasad
- Department of Hepatobiliary and Transplant Surgery, St. James’s University Hospital, Leeds LS9 7TF, UK;
| | - Nicola Ingram
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Leeds LS9 7TF, UK; (I.W.); (L.E.M.); (A.C.); (A.A.); (I.M.C.); (A.N.); (N.I.)
| |
Collapse
|
16
|
Eisenblätter M, Wildgruber M. Optical and Optoacoustic Imaging Probes. Recent Results Cancer Res 2020; 216:337-355. [PMID: 32594392 DOI: 10.1007/978-3-030-42618-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Tissue has characteristic properties when it comes to light absorption and scattering. For optical (OI) and optoacoustic imaging (OAI) these properties can be utilised to visualise biological tissue characteristics, as, for example, the oxygenation state of haemoglobin alters the optical and optoacoustic properties of the molecule.
Collapse
Affiliation(s)
- Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Freiburg im Breisgau, Germany.
| | - Moritz Wildgruber
- Department of Radiology, Ludwig Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
17
|
Langer NH, Langer SW, Johannesen HH, Hansen AE, Costa J, Klausen TL, Forman J, Olin A, Rasmussen SH, Sørensen JB, Löfgren J, Kjær A, Fischer BM. Very Early Response Evaluation by PET/MR in Patients with Lung Cancer-Timing and Feasibility. Diagnostics (Basel) 2019; 9:diagnostics9010035. [PMID: 30917539 PMCID: PMC6468790 DOI: 10.3390/diagnostics9010035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 12/25/2022] Open
Abstract
Purpose: With the increasing number of therapy options available for patients with lung cancer, early response evaluation is needed. We performed this pilot study to assess the feasibility of early, repeated Positron emission tomography-magnetic resonance (PET/MR), the impact of timing and the capability for response prediction in lung tumors during chemotherapy. Methods: Patients with stage IV non-small cell lung cancer referred for chemotherapy were prospectively recruited. Fluorine-18-Fluorodeoxyglucose(18F-FDG)-PET/MR scans were performed prior to, during and after the first or second cycle of chemotherapy. Primary tumors were defined on all scans and size, FDG-uptake and apparent diffusion coefficient (ADC) were measured. Early response was described over time and a Standard Linear Mixed Model was applied to analyze changes over time. Results: 45 FDG-PET/MR scans were performed in 11 patients. Whereas the overall changes measured by ADC did not change significantly, there was an overall significant decrease in FDG-uptake from pre to post treatment scans. There was no difference in the FDG-uptake measured 1 or 3 weeks after therapy, but uptake measured 2 weeks after therapy differed from measurements at week 3. Changes measured in patients scanned during the first treatment cycle appeared more pronounced than during the second cycle. Conclusions: This pilot study indicates that response evaluation shortly after initiation of chemotherapy appears concordant with later evaluation and probably more reliable than evaluation midway between cycles. Responses during or after the first cycle of chemotherapy rather than during subsequent cycles are likely to be more readily measured.
Collapse
Affiliation(s)
- Natasha Hemicke Langer
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Seppo W Langer
- Dept. of Oncology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Helle Hjorth Johannesen
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Adam Espe Hansen
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Junia Costa
- Dept. of Radiology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Thomas Levin Klausen
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Julie Forman
- Section of Biostatistics, Dept. of Public Health, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Anders Olin
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Sine Hvid Rasmussen
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Jens Benn Sørensen
- Dept. of Oncology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Johan Löfgren
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Andreas Kjær
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Barbara Malene Fischer
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
- PET Centre, School of Biomedical Engineering and Imaging Sciences, Kings College Hospital, Guy's & St Thomas Hospital, SE1 9RT London, UK.
| |
Collapse
|
18
|
Gozzi M, Schwarze B, Hey-Hawkins E. Half- and mixed-sandwich metallacarboranes for potential applications in medicine. PURE APPL CHEM 2019. [DOI: 10.1515/pac-2018-0806] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
Today, medicinal chemistry is still clearly dominated by organic chemistry, and commercially available boron-based drugs are rare. In contrast to hydrocarbons, boranes prefer the formation of polyhedral clusters via delocalized 3c2e bonds, such as polyhedral dicarba-closo-dodecaborane(12) (closo-C2B10H12). These clusters have remarkable biological stability, and the three isomers, 1,2- (ortho), 1,7- (meta), and 1,12-dicarba-closo-dodecaborane(12) (para), have attracted much interest due to their unique structural features. Furthermore, anionic nido clusters ([7,8-C2B9H11]2−), derived from the neutral icosahedral closo cluster 1,2-dicarba-closo-dodecaborane(12) by deboronation followed by deprotonation are suitable ligands for transition metals and offer the possibility to form metallacarboranes, for example via coordination through the upper pentagonal face of the cluster. The isolobal analogy between the cyclopentadienyl(–1) ligand (Cp−) and [C2B9H11]2− clusters (dicarbollide anion, Cb2−) is the motivation in using Cb2− as ligand for coordination to a metal center to design compounds for various applications. This review focuses on potential applications of half- and mixed-sandwich-type transition metal complexes in medicine.
Collapse
Affiliation(s)
- Marta Gozzi
- Universität Leipzig, Institut für Anorganische Chemie , Johannisallee 29 , 04103 Leipzig , Germany
| | - Benedikt Schwarze
- Universität Leipzig, Institut für Anorganische Chemie , Johannisallee 29 , 04103 Leipzig , Germany
| | - Evamarie Hey-Hawkins
- Universität Leipzig, Institut für Anorganische Chemie , Johannisallee 29 , 04103 Leipzig , Germany , Phone: +49-341-9736151, Fax: +49-341-9739319
| |
Collapse
|
19
|
Zhang C, Ling X, Guo Y, Yuan C, Cheng H, Ye X, Ma R, Zhang Y, Li Y, Chang X, Kong B, Liu T, Cui H. Evaluation of COC183B2 antibody targeting ovarian cancer by near-infrared fluorescence imaging. Chin J Cancer Res 2019; 31:673-685. [PMID: 31564810 PMCID: PMC6736662 DOI: 10.21147/j.issn.1000-9604.2019.04.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective To evaluate the imaging potential of a novel near-infrared (NIR) probe conjugated to COC183B2 monoclonal antibodies (MAb) in ovarian cancer (OC). Methods The expression of OC183B2 antigen in OC was determined by immunohistochemical (IHC) staining using tissue microarrays with the H-score system and immunofluorescence (IF) staining of tumor cell lines. Imaging probes with the NIR fluorescent dye cyanine 7 (Cy7) conjugated to COC183B2 Mab were chemically engineered. OC183B2-positive human OC cells (SKOV3-Luc) were injected subcutaneously into BALB/c nude mice. Bioluminescent imaging (BLI) was performed to detect tumor location and growth. COC183B2-Cy7 at 1.1, 3.3, 10, or 30 μg were used for in vivo fluorescence imaging, and phosphate-buffered saline (PBS), free Cy7 dye and mouse isotype immunoglobulin G (IgG)-Cy7 (delivered at the same doses as COC183B2-Cy7) were used as controls. Results The expression of OC183B2 with a high H-score was more prevalent in OC tissue than fallopian tube (FT) tissue. Among 417 OC patients, the expression of OC183B2 was significantly correlated with the histological subtype, histological grade, residual tumor size, relapse state and survival status. IF staining demonstrated that COC183B2 specifically expressed in SKOV3 cells but not HeLa cells. In vivo NIR fluorescence imaging indicated that COC183B2-Cy7 was mainly distributed in the xenograft and liver with optimal tumor-to-background (T/B) ratios in the xenograft at 30 μg dose. The highest fluorescent signals in the tumor were observed at 96 h post-injection (hpi). Ex vivo fluorescence imaging revealed the fluorescent signals mainly from the tumor and liver. IHC analysis confirmed that xenografts were OC183B2 positive. Conclusions COC183B2 is a good candidate for NIR fluorescence imaging and imaging-guided surgery in OC.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Xinyu Ling
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yanxiu Guo
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Cunzhong Yuan
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Hongyan Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Xue Ye
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Ruiqiong Ma
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, Beijing 100044, China
| | - Yi Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| | - Xiaohong Chang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Tao Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Heng Cui
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China.,Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
20
|
Asano A, Ueda S, Kuji I, Yamane T, Takeuchi H, Hirokawa E, Sugitani I, Shimada H, Hasebe T, Osaki A, Saeki T. Intracellular hypoxia measured by 18F-fluoromisonidazole positron emission tomography has prognostic impact in patients with estrogen receptor-positive breast cancer. Breast Cancer Res 2018; 20:78. [PMID: 30053906 PMCID: PMC6063018 DOI: 10.1186/s13058-018-0970-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/20/2018] [Indexed: 02/08/2023] Open
Abstract
Background Hypoxia is a key driver of cancer progression. We evaluated the prognostic impact of 18F-fluoromisonidazole (FMISO) prior to treatment in patients with breast cancer. Methods Forty-four patients with stage II/III primary breast cancer underwent positron emission tomography/computed with 18F-fluorodeoxyglucose (FDG-PET/CT) and FMISO. After measurement by FDG-PET/CT, the tissue-to-blood ratio (TBR) was obtained using FMISO-PET/CT. FMISO-TBR was compared for correlation with clinicopathological factors, disease-free survival (DFS), and overall survival (OS). Multiplex cytokines were analyzed for the correlation of FMISO-TBR. Results Tumors with higher nuclear grade and negativities of estrogen receptor (ER) and progesterone receptor had significantly higher FMISO-TBR than other tumors. Kaplan-Meier survival curves showed that patients with a higher FMISO-TBR (cutoff, 1.48) had a poorer prognosis of DFS (p = 0.0007) and OS (p = 0.04) than those with a lower FMISO-TBR. Multivariate analysis indicated that higher FMISO-TBR and ER negativity were independent predictors of shorter DFS (p = 0.01 and 0.03). Higher FMISO-TBR was associated with higher plasma levels of angiogenic hypoxic markers such as vascular endothelial growth factor, transforming growth factor-α, and interleukin 8. Conclusions FMISO-PET/CT is useful for assessing the prognosis of patients with breast cancer, but it should be stratified by ER status. Trial registration UMIN Clinical Trials Registry, UMIN000006802. Registered on 1 December 2011. Electronic supplementary material The online version of this article (10.1186/s13058-018-0970-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aya Asano
- Department of Breast Oncology, Saitama Medical University, 38 Morohongo, Moroyama-machi, Irumagun, Saitama, 350-0451, Japan
| | - Shigeto Ueda
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan.
| | - Tomohiko Yamane
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Hideki Takeuchi
- Department of Breast Oncology, Saitama Medical University, 38 Morohongo, Moroyama-machi, Irumagun, Saitama, 350-0451, Japan
| | - Eiko Hirokawa
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Ikuko Sugitani
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Hiroko Shimada
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Takahiro Hasebe
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Akihiko Osaki
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | - Toshiaki Saeki
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| |
Collapse
|
21
|
Mankoff DA, Katz SI. PET imaging for assessing tumor response to therapy. J Surg Oncol 2018; 118:362-373. [PMID: 29938396 DOI: 10.1002/jso.25114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Abstract
Positron emission tomography (PET) is a radioisotope imaging technique capable of quantifying the regional distribution of molecular imaging probes targeted to biochemical pathways and processes allowing direct measurement of biochemical changes induced by cancer therapy, including the activity of targeted growth pathways and cellular populations. In this manuscript, we review the underlying principles of PET imaging, choices for PET radiopharmaceuticals, methods for tumor analysis and PET applications for cancer therapy response assessment including potential future directions.
Collapse
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sharyn I Katz
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Neumaier CE, Baio G, Ferrini S, Corte G, Daga A. MR and Iron Magnetic Nanoparticles. Imaging Opportunities in Preclinical and Translational Research. TUMORI JOURNAL 2018; 94:226-33. [DOI: 10.1177/030089160809400215] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ultrasmall superparamagnetic iron oxide nanoparticles and magnetic resonance imaging provide a non-invasive method to detect and label tumor cells. These nanoparticles exhibit unique properties of superparamagnetism and can be utilized as excellent probes for magnetic resonance imaging. Most work has been performed using a magnetic resonance scanner with high field strength up to 7 T. Ultrasmall superparamagnetic iron oxide nanoparticles may represent a suitable tool for labeling molecular probes that target specific tumor-associated markers for in vitro and in vivo detection by magnetic resonance imaging. In our study, we demonstrated that magnetic resonance imaging at 1.5 T allows the detection of ultrasmall superparamagnetic iron oxide nanoparticle conjugated antibody specifically bound to human tumor cells in vitro and in vivo, and that the magnetic resonance signal intensity correlates with the concentration of ultrasmall superparamagnetic iron oxide nanoparticle antibody used and with the antigen density at the cell surface. The experiments were performed using two different means of targeting: direct and indirect magnetic tumor targeting. The imaging of tumor antigens using immunospecific contrast agents is a rapidly evolving field, which can potentially aid in early disease detection, monitoring of treatment efficacy, and drug development. Cell labeling by iron oxide nanoparticles has emerged as a potentially powerful tool to monitor trafficking of a large number of cells in the cell therapy field. We also studied the labeling of natural killer cells with iron nanoparticles to a level that would allow the detection of their signal intensity with a clinical magnetic resonance scanner at 1.5 T. Magnetic resonance imaging and iron magnetic nanoparticles are able to increase the accuracy and the specificity of imaging and represent new imaging opportunities in preclinical and translational research.
Collapse
Affiliation(s)
- Carlo Emanuele Neumaier
- Department of Diagnostic Imaging, Istituto Nazionale per la Ricerca sul Cancro, IST, Genoa, Italy
| | - Gabriella Baio
- Department of Diagnostic Imaging, Istituto Nazionale per la Ricerca sul Cancro, IST, Genoa, Italy
| | - Silvano Ferrini
- Laboratory of Immunological Therapy, Istituto Nazionale per la Ricerca sul Cancro, IST, Genoa, Italy
| | - Giorgio Corte
- Translational Oncology, Istituto Nazionale per la Ricerca sul Cancro, IST, Genoa, Italy
| | - Antonio Daga
- Translational Oncology, Istituto Nazionale per la Ricerca sul Cancro, IST, Genoa, Italy
| |
Collapse
|
23
|
Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, Barker AD, Bastani R, Bhatia S, Bluestone JA, Brawley O, Butte AJ, Coit DG, Davidson NE, Davis M, DePinho RA, Diasio RB, Draetta G, Frazier AL, Futreal A, Gambhir SS, Ganz PA, Garraway L, Gerson S, Gupta S, Heath J, Hoffman RI, Hudis C, Hughes-Halbert C, Ibrahim R, Jadvar H, Kavanagh B, Kittles R, Le QT, Lippman SM, Mankoff D, Mardis ER, Mayer DK, McMasters K, Meropol NJ, Mitchell B, Naredi P, Ornish D, Pawlik TM, Peppercorn J, Pomper MG, Raghavan D, Ritchie C, Schwarz SW, Sullivan R, Wahl R, Wolchok JD, Wong SL, Yung A. Future cancer research priorities in the USA: a Lancet Oncology Commission. Lancet Oncol 2017; 18:e653-e706. [PMID: 29208398 PMCID: PMC6178838 DOI: 10.1016/s1470-2045(17)30698-8] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022]
Abstract
We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US$2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.
Collapse
Affiliation(s)
| | - Chi Van Dang
- Ludwig Institute for Cancer Research New York, NY; Wistar Institute, Philadelphia, PA, USA.
| | - David B Agus
- University of Southern California, Beverly Hills, CA, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Alan Ashworth
- University of California San Francisco, San Francisco, CA, USA
| | | | - Roshan Bastani
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Sangeeta Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey A Bluestone
- University of California San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Atul J Butte
- University of California San Francisco, San Francisco, CA, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Mark Davis
- California Institute for Technology, Pasadena, CA, USA
| | | | | | - Giulio Draetta
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Lindsay Frazier
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Patricia A Ganz
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Levi Garraway
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA; Eli Lilly and Company, Boston, MA, USA
| | | | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Faculty of Medicine and IHPME, University of Toronto, Toronto, Canada
| | - James Heath
- California Institute for Technology, Pasadena, CA, USA
| | - Ruth I Hoffman
- American Childhood Cancer Organization, Beltsville, MD, USA
| | - Cliff Hudis
- Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Chanita Hughes-Halbert
- Medical University of South Carolina and the Hollings Cancer Center, Charleston, SC, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Hossein Jadvar
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Kavanagh
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| | - Rick Kittles
- College of Medicine, University of Arizona, Tucson, AZ, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | - Scott M Lippman
- University of California San Diego Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - David Mankoff
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Mardis
- The Institute for Genomic Medicine at Nationwide Children's Hospital Columbus, OH, USA; College of Medicine, Ohio State University, Columbus, OH, USA
| | - Deborah K Mayer
- University of North Carolina Lineberger Cancer Center, Chapel Hill, NC, USA
| | - Kelly McMasters
- The Hiram C Polk Jr MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dean Ornish
- University of California San Francisco, San Francisco, CA, USA
| | - Timothy M Pawlik
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | | | - Martin G Pomper
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Raghavan
- Levine Cancer Institute, Carolinas HealthCare, Charlotte, NC, USA
| | | | - Sally W Schwarz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Richard Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Sandra L Wong
- Department of Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alfred Yung
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Zhou Z, Lu ZR. Molecular imaging of the tumor microenvironment. Adv Drug Deliv Rev 2017; 113:24-48. [PMID: 27497513 DOI: 10.1016/j.addr.2016.07.012] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment plays a critical role in tumor initiation, progression, metastasis, and resistance to therapy. It is different from normal tissue in the extracellular matrix, vascular and lymphatic networks, as well as physiologic conditions. Molecular imaging of the tumor microenvironment provides a better understanding of its function in cancer biology, and thus allowing for the design of new diagnostics and therapeutics for early cancer diagnosis and treatment. The clinical translation of cancer molecular imaging is often hampered by the high cost of commercialization of targeted imaging agents as well as the limited clinical applications and small market size of some of the agents. Because many different cancer types share similar tumor microenvironment features, the ability to target these biomarkers has the potential to provide clinically translatable molecular imaging technologies for a spectrum of cancers and broad clinical applications. There has been significant progress in targeting the tumor microenvironment for cancer molecular imaging. In this review, we summarize the principles and strategies of recent advances made in molecular imaging of the tumor microenvironment, using various imaging modalities for early detection and diagnosis of cancer.
Collapse
|
25
|
Eary JF. Diagnostic Applications of Nuclear Medicine: Sarcomas. NUCLEAR ONCOLOGY 2017:1047-1064. [DOI: 10.1007/978-3-319-26236-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Kimbrough CW, Khanal A, Zeiderman M, Khanal BR, Burton NC, McMasters KM, Vickers SM, Grizzle WE, McNally LR. Targeting Acidity in Pancreatic Adenocarcinoma: Multispectral Optoacoustic Tomography Detects pH-Low Insertion Peptide Probes In Vivo. Clin Cancer Res 2015; 21:4576-85. [PMID: 26124201 PMCID: PMC4609270 DOI: 10.1158/1078-0432.ccr-15-0314] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/12/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND pH-low insertion peptides (pHLIP) can serve as a targeting moiety that enables pH-sensitive probes to detect solid tumors. Using these probes in conjunction with multispectral optoacoustic tomography (MSOT) is a promising approach to improve imaging for pancreatic cancer. METHODS A pH-sensitive pHLIP (V7) was conjugated to 750 NIR fluorescent dye and evaluated as a targeted probe for pancreatic adenocarcinoma. The pH-insensitive K7 pHLIP served as an untargeted control. Probe binding was assessed in vitro at pH 7.4, 6.8, and 6.6 using human pancreatic cell lines S2VP10 and S2013. Using MSOT, semiquantitative probe accumulation was then assessed in vivo with a murine orthotopic pancreatic adenocarcinoma model. RESULTS In vitro, the V7-750 probe demonstrated significantly higher fluorescence at pH 6.6 compared with pH 7.4 (S2VP10, P = 0.0119; S2013, P = 0.0160), whereas no difference was observed with the K7-750 control (S2VP10, P = 0.8783; S2013, P = 0.921). In the in vivo S2VP10 model, V7-750 probe resulted in 782.5 MSOT a.u. signal compared with 5.3 MSOT a.u. in K7-750 control in tumor (P = 0.0001). Similarly, V7-750 probe signal was 578.3 MSOT a.u. in the S2013 model compared with K7-750 signal at 5.1 MSOT a.u. (P = 0.0005). There was minimal off-target accumulation of the V7-750 probe within the liver or kidney, and probe distribution was confirmed with ex vivo imaging. CONCLUSIONS Compared with pH-insensitive controls, V7-750 pH-sensitive probe specifically targets pancreatic adenocarcinoma and has minimal off-target accumulation. The noninvasive detection of pH-targeted probes by means of MSOT represents a promising modality to improve the detection and monitoring of pancreatic cancer.
Collapse
Affiliation(s)
- Charles W Kimbrough
- The Hiram C. Polk Jr, MD Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Anil Khanal
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Matthew Zeiderman
- The Hiram C. Polk Jr, MD Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Bigya R Khanal
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | | | - Kelly M McMasters
- The Hiram C. Polk Jr, MD Department of Surgery, University of Louisville, Louisville, Kentucky
| | | | | | - Lacey R McNally
- Department of Medicine, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
27
|
Paar S, Umathum R, Jiang X, Majer CL, Peter J. Development and investigation of a magnetic resonance imaging-compatible microlens-based optical detector. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:095007. [PMID: 26359811 DOI: 10.1117/1.jbo.20.9.095007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/07/2015] [Indexed: 06/05/2023]
Abstract
A noncontact optical detector for in vivo imaging has been developed that is compatible with magnetic resonance imaging (MRI). The optical detector employs microlens arrays and might be classified as a plenoptic camera. As a resulting of its design, the detector possesses a slim thickness and is self-shielding against radio frequency (RF) pulses. For experimental investigation, a total of six optical detectors were arranged in a cylindrical fashion, with the imaged object positioned in the center of this assembly. A purposely designed RF volume resonator coil has been developed and is incorporated within the optical imaging system. The whole assembly was placed into the bore of a 1.5 T patient-sized MRI scanner. Simple-geometry phantom studies were performed to assess compatibility and performance characteristics regarding both optical and MR imaging systems. A bimodal ex vivo nude mouse measurement was conducted. From the MRI data, the subject surface was extracted. Optical images were projected on this surface by means of an inverse mapping algorithm. Simultaneous measurements did not reveal influences from the magnetic field and RF pulses onto optical detector performance (spatial resolution, sensitivity). No significant influence of the optical imaging system onto MRI performance was detectable.
Collapse
|
28
|
Zhou B, Wang H, Liu R, Wang M, Deng H, Giglio BC, Gill PS, Shan H, Li Z. PET Imaging of Dll4 Expression in Glioblastoma and Colorectal Cancer Xenografts Using (64)Cu-Labeled Monoclonal Antibody 61B. Mol Pharm 2015; 12:3527-34. [PMID: 26288060 DOI: 10.1021/acs.molpharmaceut.5b00105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Delta-like ligand 4 (Dll4) expressed in tumor cells plays a key role to promote tumor growth of numerous cancer types. Based on a novel antihuman Dll4 monoclonal antibody (61B), we developed a (64)Cu-labeled probe for positron emission tomography (PET) imaging of tumor Dll4 expression. In this study, 61B was conjugated with the (64)Cu-chelator DOTA through lysine on the antibody. Human IgG (hIgG)-DOTA, which did not bind to Dll4, was also prepared as a control. The Dll4 binding activity of the probes was evaluated through the bead-based binding assay with Dll4-alkaline phosphatase. The resulting PET probes were evaluated in U87MG glioblastoma and HT29 colorectal cancer xenografts in athymic nude mice. Our results demonstrated that the 61B-DOTA retained (77.2 ± 3.7) % Dll4 binding activity of the unmodified 61B, which is significantly higher than that of hIgG-DOTA (0.06 ± 0.03) %. Confocal microscopy analysis confirmed that 61B-Cy5.5, but not IgG-Cy5.5, predominantly located within the U87MG and HT29 cells cytoplasm. U87MG cells showed higher 61B-Cy5.5 binding as compared to HT29 cells. In U87MG xenografts, 61B-DOTA-(64)Cu demonstrated remarkable tumor accumulation (10.5 ± 1.7 and 10.2 ± 1.2%ID/g at 24 and 48 h postinjection, respectively). In HT29 xenografts, tumor accumulation of 61B-DOTA-(64)Cu was significantly lower than that of U87MG (7.3 ± 1.3 and 6.6 ± 1.3%ID/g at 24 and 48 h postinjection, respectively). The tumor accumulation of 61B-DOTA-(64)Cu was significantly higher than that of hIgG-DOTA-(64)Cu in both xenografts models. Immunofluorescence staining of the tumor tissues further confirmed that tumor accumulation of 61B-Cy5.5 was correlated well with in vivo PET imaging data using 61B-DOTA-(64)Cu. In conclusion, 61B-DOTA-(64)Cu PET probe was successfully synthesized and demonstrated prominent tumor uptake by targeting Dll4. 61B-DOTA-(64)Cu has great potential to be used for noninvasive Dll4 imaging, which could be valuable for tumor detection, Dll4 expression level evaluation, and Dll4-based treatment monitoring.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou 510630, China.,Biomedical Research Imaging Center, Department of Radiology, University of North Carolina , Chapel Hill, North Carolina 27514, United States
| | - Hui Wang
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina , Chapel Hill, North Carolina 27514, United States
| | - Ren Liu
- Department of Pathology, University of Southern California , Los Angeles, California 90033, United States
| | - Mengzhe Wang
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina , Chapel Hill, North Carolina 27514, United States
| | - Huaifu Deng
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina , Chapel Hill, North Carolina 27514, United States
| | - Benjamin C Giglio
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina , Chapel Hill, North Carolina 27514, United States
| | - Parkash S Gill
- Department of Pathology, University of Southern California , Los Angeles, California 90033, United States
| | - Hong Shan
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou 510630, China.,Interventional Radiology Institute, Sun Yat-sen University , Guangzhou 510630, China
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina , Chapel Hill, North Carolina 27514, United States
| |
Collapse
|
29
|
Fudala R, Raut S, Maliwal BP, Zerda TW, Gryczynski I, Simanek E, Borejdo J, Rich R, Akopova I, Gryczynski Z. FRET enhanced fluorescent nanodiamonds. Curr Pharm Biotechnol 2015; 14:1127-33. [PMID: 22394126 DOI: 10.2174/138920101413140605110711] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 11/09/2011] [Accepted: 12/10/2011] [Indexed: 11/22/2022]
Abstract
Fluorescent nanodiamonds (FNDs) are one of the new and very promising biocompatible nanomaterials that can be used both as a fluorescence imaging agent and a highly versatile platform for controlled functionalization to target and deliver a wide spectrum of therapeutic agents. Among the remarkable fluorescence properties are excellent photostability, emission between 600-700nm, quantum yield of 1 and moderately long fluorescence lifetimes. However the low absorption cross section of fluorescent (N-V)(-) centers limits FNDs' brightness. In this work we show that an approach based on the Forster resonance energy transfer (FRET) may significantly enhance the fluorescence signal observed from a single ND. We demonstrate that organic dyes (fluorophores) attached to the FND surface can efficiently transfer the excitation energy to (N-V)(-) centers. Multiple dyes positioned in close proximity to the ND facile surface may serve as harvesting antennas transferring excitation energy to the fluorescent centers. We propose that, with the help of some of the functional groups present on the FND surface, we can either directly link flurophores or use scalable dendrimer chemistry to position many organic dyes at a calibrated distance. Also, the remaining multiple functional groups will be still available for particle targeting and drug delivery. This opens a new way for designing a new type of theranostics particles of ultrahigh brightness, high photostability, specific targeting, and high capacity for drug delivery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zygmunt Gryczynski
- Department of Physics and Astronomy, Texas Christian University, TCU Box 298840, Fort Worth, TX 76129, USA.
| |
Collapse
|
30
|
Bannas P, Lenz A, Kunick V, Well L, Fumey W, Rissiek B, Haag F, Schmid J, Schütze K, Eichhoff A, Trepel M, Adam G, Ittrich H, Koch-Nolte F. Molecular imaging of tumors with nanobodies and antibodies: Timing and dosage are crucial factors for improvedin vivodetection. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:367-78. [DOI: 10.1002/cmmi.1637] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/23/2015] [Accepted: 02/03/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Peter Bannas
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Alexander Lenz
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Valentin Kunick
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Lennart Well
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - William Fumey
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Björn Rissiek
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Department of Neurology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Friedrich Haag
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Joanna Schmid
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Kerstin Schütze
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Anna Eichhoff
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Martin Trepel
- Department of Oncology and Hematology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Harald Ittrich
- Department of Diagnostic and Interventional Radiology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| |
Collapse
|
31
|
Honarvar H, Strand J, Perols A, Orlova A, Selvaraju RK, Karlström AE, Tolmachev V. Position for Site-Specific Attachment of a DOTA Chelator to Synthetic Affibody Molecules Has a Different Influence on the Targeting Properties of
68
Ga-Compared to
111
In-Labeled Conjugates. Mol Imaging 2014; 13. [DOI: 10.2310/7290.2014.00034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Hadis Honarvar
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Joanna Strand
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Perols
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ram Kumar Selvaraju
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Amelie Eriksson Karlström
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- From Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden; and Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
32
|
Phillips E, Penate-Medina O, Zanzonico PB, Carvajal RD, Mohan P, Ye Y, Humm J, Gönen M, Kalaigian H, Schöder H, Strauss HW, Larson SM, Wiesner U, Bradbury MS. Clinical translation of an ultrasmall inorganic optical-PET imaging nanoparticle probe. Sci Transl Med 2014; 6:260ra149. [PMID: 25355699 DOI: 10.1126/scitranslmed.3009524] [Citation(s) in RCA: 491] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A first-in-human clinical trial of ultrasmall inorganic hybrid nanoparticles, "C dots" (Cornell dots), in patients with metastatic melanoma is described for the imaging of cancer. These renally excreted silica particles were labeled with (124)I for positron emission tomography (PET) imaging and modified with cRGDY peptides for molecular targeting. (124)I-cRGDY-PEG-C dot particles are inherently fluorescent, containing the dye, Cy5, so they may be used as hybrid PET-optical imaging agents for lesion detection, cancer staging, and treatment management in humans. However, the clinical translation of nanoparticle probes, including quantum dots, has not kept pace with the accelerated growth in minimally invasive surgical tools that rely on optical imaging agents. The safety, pharmacokinetics, clearance properties, and radiation dosimetry of (124)I-cRGDY-PEG-C dots were assessed by serial PET and computerized tomography after intravenous administration in patients. Metabolic profiles and laboratory tests of blood and urine specimens, obtained before and after particle injection, were monitored over a 2-week interval. Findings are consistent with a well-tolerated inorganic particle tracer exhibiting in vivo stability and distinct, reproducible pharmacokinetic signatures defined by renal excretion. No toxic or adverse events attributable to the particles were observed. Coupled with preferential uptake and localization of the probe at sites of disease, these first-in-human results suggest safe use of these particles in human cancer diagnostics.
Collapse
Affiliation(s)
- Evan Phillips
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Oula Penate-Medina
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Pat B Zanzonico
- Department of Medical Physics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Richard D Carvajal
- Department of Medicine, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Pauliah Mohan
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Yunpeng Ye
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - John Humm
- Department of Medical Physics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Hovanes Kalaigian
- Department of Medical Physics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Heiko Schöder
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - H William Strauss
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Steven M Larson
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Ulrich Wiesner
- Department of Materials Science and Engineering, Cornell University, 330 Bard Hall, Ithaca, NY 14853, USA
| | - Michelle S Bradbury
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA.
| |
Collapse
|
33
|
Su YC, Cheng TC, Leu YL, Roffler SR, Wang JY, Chuang CH, Kao CH, Chen KC, Wang HE, Cheng TL. PET imaging of β-glucuronidase activity by an activity-based 124I-trapping probe for the personalized glucuronide prodrug targeted therapy. Mol Cancer Ther 2014; 13:2852-63. [PMID: 25277385 DOI: 10.1158/1535-7163.mct-14-0212] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Beta-glucuronidase (βG) is a potential biomarker for cancer diagnosis and prodrug therapy. The ability to image βG activity in patients would assist in personalized glucuronide prodrug cancer therapy. However, whole-body imaging of βG activity for medical usage is not yet available. Here, we developed a radioactive βG activity-based trapping probe for positron emission tomography (PET). We generated a (124)I-tyramine-conjugated difluoromethylphenol beta-glucuronide probe (TrapG) to form (124)I-TrapG that could be selectively activated by βG for subsequent attachment of (124)I-tyramine to nucleophilic moieties near βG-expressing sites. We estimated the specificity of a fluorescent FITC-TrapG, the cytotoxicity of tyramine-TrapG, and the serum half-life of (124)I-TrapG. βG targeting of (124)I-TrapG in vivo was examined by micro-PET. The biodistribution of (131)I-TrapG was investigated in different organs. Finally, we imaged the endogenous βG activity and assessed its correlation with therapeutic efficacy of 9-aminocamptothecin glucuronide (9ACG) prodrug in native tumors. FITC-TrapG showed specific trapping at βG-expressing CT26 (CT26/mβG) cells but not in CT26 cells. The native TrapG probe possessed low cytotoxicity. (124)I-TrapG preferentially accumulated in CT26/mβG but not CT26 cells. Meanwhile, micro-PET and whole-body autoradiography results demonstrated that (124)I-TrapG signals in CT26/mβG tumors were 141.4-fold greater than in CT26 tumors. Importantly, Colo205 xenografts in nude mice that express elevated endogenous βG can be monitored by using infrared glucuronide trapping probes (NIR-TrapG) and suppressed by 9ACG prodrug treatment. (124)I-TrapG exhibited low cytotoxicity allowing long-term monitoring of βG activity in vivo to aid in the optimization of prodrug targeted therapy.
Collapse
Affiliation(s)
- Yu-Cheng Su
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan. Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ta-Chun Cheng
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ling Leu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jaw-Yuan Wang
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hung Chuang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Han Kao
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kai-Chuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.
| | - Tian-Lu Cheng
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan. Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan. Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
34
|
Iwaki S, Hokamura K, Ogawa M, Takehara Y, Muramatsu Y, Yamane T, Hirabayashi K, Morimoto Y, Hagisawa K, Nakahara K, Mineno T, Terai T, Komatsu T, Ueno T, Tamura K, Adachi Y, Hirata Y, Arita M, Arai H, Umemura K, Nagano T, Hanaoka K. A design strategy for small molecule-based targeted MRI contrast agents: their application for detection of atherosclerotic plaques. Org Biomol Chem 2014; 12:8611-8. [DOI: 10.1039/c4ob01270d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
35
|
Lütje S, Rijpkema M, Helfrich W, Oyen WJG, Boerman OC. Targeted Radionuclide and Fluorescence Dual-modality Imaging of Cancer: Preclinical Advances and Clinical Translation. Mol Imaging Biol 2014; 16:747-55. [DOI: 10.1007/s11307-014-0747-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Biological evaluation of 131I- and CF750-labeled Dmab(scFv)-Fc antibodies for xenograft imaging of CD25-positive tumors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:459676. [PMID: 24864244 PMCID: PMC4017786 DOI: 10.1155/2014/459676] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/17/2014] [Accepted: 03/17/2014] [Indexed: 02/05/2023]
Abstract
A Dmab(scFv)-Fc antibody containing the single chain variable fragment of a humanized daclizumab antibody and the Fc fragment of a human IgG1 antibody was produced via recombinant expression in Pichia pastoris. The Dmab(scFv)-Fc antibody forms a dimer in solution, and it specifically binds CD25-positive tumor cells and tumor tissues. For tumor imaging, the Dmab(scFv)-Fc antibody was labeled with the 131I isotope and CF750 fluorescent dye, respectively. After intravenous injection of mice bearing CD25-positive tumor xenografts, tumor uptake of the (131)I-Dmab(scFv)-Fc antibody was visible at 1 h, and clear images were obtained at 5 h using SPECT/CT. After systemic administration of the CF750-Dmab(scFv)-Fc antibody, tumor uptake was present as early as 1 h, and tumor xenografts could be kinetically imaged within 9 h after injection. These results indicate that the Dmab(scFv)-Fc antibody rapidly and specifically targets CD25-positive tumor cells, suggesting the potential of this antibody as an imaging agent for the diagnosis of lymphomatous-type ATLL.
Collapse
|
37
|
Meyer JP, Probst KC, Westwell AD. Radiochemical synthesis of 2'-[18F]-labelled and 3'-[18F]-labelled nucleosides for positron emission tomography imaging. J Labelled Comp Radiopharm 2014; 57:333-7. [PMID: 24692121 DOI: 10.1002/jlcr.3197] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/10/2014] [Accepted: 02/23/2014] [Indexed: 01/11/2023]
Abstract
This review article considers 2'-labelled and 3'-labelled nucleosides, which are of great importance as positron emission tomography (PET) probes in clinical diagnostics and PET research. Although the radiochemical preparation of several [(18)F]-labelled nucleosides such as [(18)F]fluorothymidine or [(18)F](fluoroarabinofuranosyl)cytosine has been accomplished within the last two decades, a number of potentially interesting nucleoside-based biomarkers are not yet available for automated good manufacturing practice production due to the lack of fast and efficient synthetic methods for late-stage [(18)F]-introduction. In order to meet recent demands for new PET-based biomarkers in various clinical applications, appropriate precursors that can easily be fluorinated and deprotected need to be developed.
Collapse
Affiliation(s)
- Jan-Philip Meyer
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, Wales, UK; Wales Research & Diagnostic PET Imaging Centre (PETIC), Institute for Translation, Innovation, Methodology & Engagement (TIME), School of Medicine, Heath Park, Cardiff University, Cardiff, CF14 4XN, Wales, UK
| | | | | |
Collapse
|
38
|
Garofalakis A, Dubois A, Thézé B, Czarny B, Tavitian B, Ducongé F. Fusion of [(18)F]FDG PET with fluorescence diffuse optical tomography to improve validation of probes and tumor imaging. Mol Imaging Biol 2014; 15:316-25. [PMID: 22927057 PMCID: PMC3647087 DOI: 10.1007/s11307-012-0581-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Purpose Given the progress of fluorescence diffuse optical tomography (fDOT) technology, here, we study the additional benefits provided by multimodal PET/fDOT imaging by comparing the biodistribution of 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG) in tumors with three fluorescent probes: a glucose analog, a protease activatable optical probe, and a ligand of αvβ3 integrin. Procedures Sequential fDOT/PET/computed tomography (CT) imaging of mice was performed with a custom multimodal mouse support that allows the subject to be transferred between the fDOT and the PET/CT scanners. Experiments were performed in xenografted tumor models derived from the human breast cancer line MDA-MB 231 and compared to ex vivo analysis. Results The three-dimensional signals showed that the fluorescent glucose analog is not colocalized with [18F]FDG, raising questions about its use as a surrogate probe of the PET tracer. Fusion of [18F]FDG with the other fluorescent probes showed evidence of high variability both for the protease activity and the αvβ3 integrin expression during tumor growth. Conclusion The added value of hybrid PET/fDOT over the two modalities was demonstrated for cross-validation of probes and for better characterization of tumor models.
Collapse
Affiliation(s)
- Anikitos Garofalakis
- CEA, Institut d'Imagerie Biomédicale, Service Hospitalier Frédéric Joliot, Orsay Cedex, France
| | | | | | | | | | | |
Collapse
|
39
|
Automated tracking of quantitative assessments of tumor burden in clinical trials. Transl Oncol 2014; 7:23-35. [PMID: 24772204 DOI: 10.1593/tlo.13796] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/13/2014] [Accepted: 01/15/2014] [Indexed: 11/18/2022] Open
Abstract
THERE ARE TWO KEY CHALLENGES HINDERING EFFECTIVE USE OF QUANTITATIVE ASSESSMENT OF IMAGING IN CANCER RESPONSE ASSESSMENT: 1) Radiologists usually describe the cancer lesions in imaging studies subjectively and sometimes ambiguously, and 2) it is difficult to repurpose imaging data, because lesion measurements are not recorded in a format that permits machine interpretation and interoperability. We have developed a freely available software platform on the basis of open standards, the electronic Physician Annotation Device (ePAD), to tackle these challenges in two ways. First, ePAD facilitates the radiologist in carrying out cancer lesion measurements as part of routine clinical trial image interpretation workflow. Second, ePAD records all image measurements and annotations in a data format that permits repurposing image data for analyses of alternative imaging biomarkers of treatment response. To determine the impact of ePAD on radiologist efficiency in quantitative assessment of imaging studies, a radiologist evaluated computed tomography (CT) imaging studies from 20 subjects having one baseline and three consecutive follow-up imaging studies with and without ePAD. The radiologist made measurements of target lesions in each imaging study using Response Evaluation Criteria in Solid Tumors 1.1 criteria, initially with the aid of ePAD, and then after a 30-day washout period, the exams were reread without ePAD. The mean total time required to review the images and summarize measurements of target lesions was 15% (P < .039) shorter using ePAD than without using this tool. In addition, it was possible to rapidly reanalyze the images to explore lesion cross-sectional area as an alternative imaging biomarker to linear measure. We conclude that ePAD appears promising to potentially improve reader efficiency for quantitative assessment of CT examinations, and it may enable discovery of future novel image-based biomarkers of cancer treatment response.
Collapse
|
40
|
Hussain T, Nguyen QT. Molecular imaging for cancer diagnosis and surgery. Adv Drug Deliv Rev 2014; 66:90-100. [PMID: 24064465 DOI: 10.1016/j.addr.2013.09.007] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/07/2013] [Accepted: 09/13/2013] [Indexed: 12/11/2022]
Abstract
Novel molecular imaging techniques have the potential to significantly enhance the diagnostic and therapeutic approaches for cancer treatment. For solid tumors in particular, novel molecular enhancers for imaging modalities such as US, CT, MRI and PET may facilitate earlier and more accurate diagnosis and staging which are prerequisites for successful surgical therapy. Enzymatically activatable "smart" molecular MRI probes seem particularly promising because of their potential to image tumors before and after surgical removal without re-administration of the probe to evaluate completeness of surgical resection. Furthermore, the use of "smart" MR probes as part of screening programs may enable detection of small tumors throughout the body in at-risk patient populations. Dual labeling of molecular MR probes with fluorescent dyes can add real time intraoperative guidance facilitating complete tumor resection and preservation of important structures. A truly theranostic approach with the further addition of therapeutic agents to the molecular probe for adjuvant therapy is conceivable for the future.
Collapse
|
41
|
Cullinane C, Solomon B, Hicks RJ. Imaging of molecular target modulation in oncology: challenges of early clinical trials. Clin Transl Imaging 2014. [DOI: 10.1007/s40336-013-0047-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Wen PY, Kesari S, Drappatz J. Malignant gliomas: strategies to increase the effectiveness of targeted molecular treatment. Expert Rev Anticancer Ther 2014; 6:733-54. [PMID: 16759164 DOI: 10.1586/14737140.6.5.733] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, there has been increasing interest in the use of targeted molecular agents for the treatment of malignant gliomas. These agents are generally well tolerated but have demonstrated only modest activity. In this article, the current status of targeted molecular agents for malignant gliomas will be reviewed and strategies to improve their effectiveness will be discussed.
Collapse
Affiliation(s)
- Patrick Y Wen
- Harvard Medical School, Dana-Farber/Brigham and Women's Cancer Center, SW430D, Boston, MA 02115, USA.
| | | | | |
Collapse
|
43
|
Zeng MY, Wu CG, Cheng YS. Molecular imaging of inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2014; 22:3424. [DOI: 10.11569/wcjd.v22.i23.3424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
44
|
Synthesis and biological evaluation of 18F-labeled fluoropropyl tryptophan analogs as potential PET probes for tumor imaging. Eur J Med Chem 2013; 70:768-80. [DOI: 10.1016/j.ejmech.2013.10.054] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/17/2013] [Accepted: 10/21/2013] [Indexed: 11/19/2022]
|
45
|
Niikura N, Odisio BC, Tokuda Y, Symmans FW, Hortobagyi GN, Ueno NT. Latest biopsy approach for suspected metastases in patients with breast cancer. Nat Rev Clin Oncol 2013; 10:711-9. [DOI: 10.1038/nrclinonc.2013.182] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
46
|
Strand J, Honarvar H, Perols A, Orlova A, Selvaraju RK, Karlström AE, Tolmachev V. Influence of macrocyclic chelators on the targeting properties of (68)Ga-labeled synthetic affibody molecules: comparison with (111)In-labeled counterparts. PLoS One 2013; 8:e70028. [PMID: 23936372 PMCID: PMC3731330 DOI: 10.1371/journal.pone.0070028] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 06/19/2013] [Indexed: 11/26/2022] Open
Abstract
Affibody molecules are a class of small (7 kDa) non-immunoglobulin scaffold-based affinity proteins, which have demonstrated substantial potential as probes for radionuclide molecular imaging. The use of positron emission tomography (PET) would further increase the resolution and quantification accuracy of Affibody-based imaging. The rapid in vivo kinetics of Affibody molecules permit the use of the generator-produced radionuclide 68Ga (T1/2 = 67.6 min). Earlier studies have demonstrated that the chemical nature of chelators has a substantial influence on the biodistribution properties of Affibody molecules. To determine an optimal labeling approach, the macrocyclic chelators 1,4,7,10-tetraazacylododecane-1,4,7,10-tetraacetic acid (DOTA), 1,4,7-triazacyclononane-N,N,N-triacetic acid (NOTA) and 1-(1,3-carboxypropyl)-1,4,7- triazacyclononane-4,7-diacetic acid (NODAGA) were conjugated to the N-terminus of the synthetic Affibody molecule ZHER2:S1 targeting HER2. Affibody molecules were labeled with 68Ga, and their binding specificity and cellular processing were evaluated. The biodistribution of 68Ga-DOTA-ZHER2:S1,68Ga-NOTA-ZHER2:S1 and 68Ga-NODAGA-ZHER2:S1, as well as that of their 111In-labeled counterparts, was evaluated in BALB/C nu/nu mice bearing HER2-expressing SKOV3 xenografts. The tumor uptake for 68Ga-DOTA-ZHER2:S1 (17.9±0.7%IA/g) was significantly higher than for both 68Ga-NODAGA-ZHER2:S1(16.13±0.67%IA/g) and 68Ga-NOTA-ZHER2:S1 (13±3%IA/g) at 2 h after injection. 68Ga-NODAGA-ZHER2:S1 had the highest tumor-to-blood ratio (60±10) in comparison with both 68Ga-DOTA-ZHER2:S1 (28±4) and 68Ga-NOTA-ZHER2:S1 (42±11). The tumor-to-liver ratio was also higher for 68Ga-NODAGA-ZHER2:S1 (7±2) than the DOTA and NOTA conjugates (5.5±0.6 vs.3.3±0.6). The influence of chelator on the biodistribution and targeting properties was less pronounced for 68Ga than for 111In. The results of this study demonstrate that macrocyclic chelators conjugated to the N-terminus have a substantial influence on the biodistribution of HER2-targeting Affibody molecules labeled with 68Ga.This can be utilized to enhance the imaging contrast of PET imaging using Affibody molecules and improve the sensitivity of molecular imaging. The study demonstrated an appreciable difference of chelator influence for 68Ga and 111In.
Collapse
Affiliation(s)
- Joanna Strand
- Unit of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Hadis Honarvar
- Unit of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Perols
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Orlova
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ram Kumar Selvaraju
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Amelie Eriksson Karlström
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Vladimir Tolmachev
- Unit of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
47
|
Molecularly targeted agents as radiosensitizers in cancer therapy--focus on prostate cancer. Int J Mol Sci 2013; 14:14800-32. [PMID: 23863691 PMCID: PMC3742274 DOI: 10.3390/ijms140714800] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/27/2013] [Accepted: 06/27/2013] [Indexed: 12/12/2022] Open
Abstract
As our understanding of the molecular pathways driving tumorigenesis improves and more druggable targets are identified, we have witnessed a concomitant increase in the development and production of novel molecularly targeted agents. Radiotherapy is commonly used in the treatment of various malignancies with a prominent role in the care of prostate cancer patients, and efforts to improve the therapeutic ratio of radiation by technologic and pharmacologic means have led to important advances in cancer care. One promising approach is to combine molecularly targeted systemic agents with radiotherapy to improve tumor response rates and likelihood of durable control. This review first explores the limitations of preclinical studies as well as barriers to successful implementation of clinical trials with radiosensitizers. Special considerations related to and recommendations for the design of preclinical studies and clinical trials involving molecularly targeted agents combined with radiotherapy are provided. We then apply these concepts by reviewing a representative set of targeted therapies that show promise as radiosensitizers in the treatment of prostate cancer.
Collapse
|
48
|
Shi J, Cui L, Jia B, Liu Z, He P, Dong C, Jin X, Zhao H, Li F, Wang F. Technetium 99m–Labeled VQ Peptide: A New Imaging Agent for the Early Detection of Tumors or Premalignancies. Mol Imaging 2013. [DOI: 10.2310/7290.2012.00047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Jiyun Shi
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Liyang Cui
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Bing Jia
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Zhaofei Liu
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Peng He
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Chengyan Dong
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Xiaona Jin
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Huiyun Zhao
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Fang Li
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Fan Wang
- From the Medical Isotopes Research Center, Medical and Healthy Analytical Center, and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, China; and the Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
49
|
Uppal T, Bhupathiraju NDK, Vicente MGH. Synthesis and cellular properties of Near-IR BODIPY–PEG and carbohydrate conjugates. Tetrahedron 2013. [DOI: 10.1016/j.tet.2013.03.082] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
50
|
Abstract
Nanotheranostics, the integration of diagnostic and therapeutic function in one system using the benefits of nanotechnology, is extremely attractive for personalized medicine. Because treating cancer is not a one-size-fits-all scenario, it requires therapy to be adapted to the patient's specific biomolecules. Personalized and precision medicine (PM) does just that. It identifies biomarkers to gain an understanding of the diagnosis and in turn treating the specific disorder based on the precise diagnosis. By predominantly utilizing the unique properties of nanoparticles to achieve biomarker identification and drug delivery, nanotheranostics can be applied to noninvasively discover and target image biomarkers and further deliver treatment based on the biomarker distribution. This is a large and hopeful role theranostics must fill. However, as described in this expert opinion, current nanotechnology-based theranostics systems engineered for PM applications are not yet sufficient. PM is an ever-growing field that will be a driving force for future discoveries in biomedicine, especially cancer theranostics. In this article, the authors dissect the requirements for successful nanotheranostics-based PM.
Collapse
Affiliation(s)
- Tae Hyung Kim
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Seulki Lee
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, MD, USA
| |
Collapse
|